Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Thomas L. Brown is active.

Publication


Featured researches published by Thomas L. Brown.


The EMBO Journal | 1999

TGF-Beta Induces Fibronectin Synthesis Through a c-Jun N-terminal Kinase-Dependent, Smad4-Independent Pathway

Barbara A. Hocevar; Thomas L. Brown; Philip H. Howe

Transforming growth factor‐β (TGF‐β) exerts its effects on cell proliferation, differentiation and migration in part through its modulation of extracellular matrix components, such as fibronectin and plasminogen activator inhibitor‐1 (PAI‐1). Although the SMAD family of proteins recently has been shown to be a key participant in TGF‐β signaling, other signaling pathways have also been shown to be activated by TGF‐β. We report here that c‐Jun N‐terminal kinase (JNK), a member of the MAP kinase family, is activated in response to TGF‐β in the human fibrosarcoma HT1080‐derived cell line BAHgpt. Stable expression of dominant‐negative forms of JNK1 and MKK4, an upstream activator of JNK, results in loss of TGF‐β‐stimulated fibronectin mRNA and protein induction, while having little effect on TGF‐β‐induced levels of PAI‐1. The human fibronectin promoter contains three CRE elements, one of which has been shown to bind a c‐Jun–ATF‐2 heterodimer. Utilizing a GAL4 fusion trans‐reporting system, we demonstrate a decrease in transactivating potential of GAL4–c‐Jun and GAL4–ATF‐2 in dominant‐negative JNK1‐ and MKK4‐expressing cells. Finally, we show that TGF‐β‐induced fibronectin synthesis is independent of Smad4. These results demonstrate that TGF‐β‐mediated fibronectin induction requires activation of JNK which in turn modulates the activity of c‐Jun and ATF‐2 in a Smad4independent manner.


The EMBO Journal | 1997

Interferon action and apoptosis are defective in mice devoid of 2′,5′-oligoadenylate-dependent RNase L

Aimin Zhou; Jayashree M. Paranjape; Thomas L. Brown; Huiqin Nie; Sharon Naik; Beihua Dong; Ansi Chang; Bruce D. Trapp; Robert L. Fairchild; Clemencia Colmenares; Robert H. Silverman

2′,5′‐Oligoadenylate‐dependent RNase L functions in the interferon‐inducible, RNA decay pathway known as the 2‐5A system. To determine the physiological roles of the 2‐5A system, mice were generated with a targeted disruption of the RNase L gene. The antiviral effect of interferon α was impaired in RNase L−/− mice providing the first evidence that the 2‐5A system functions as an antiviral pathway in animals. In addition, remarkably enlarged thymuses in the RNase L−/− mice resulted from a suppression of apoptosis. There was a 2‐fold decrease in apoptosis in vivo in the thymuses and spleens of RNase L−/− mice. Furthermore, apoptosis was substantially suppressed in RNase L−/− thymocytes and fibroblasts treated with different apoptotic agents. These results suggest that both interferon action and apoptosis can be controlled at the level of RNA stability by RNase L. Another implication is that the 2‐5A system is likely to contribute to the antiviral activity of interferon by inducing apoptosis of infected cells.


Apoptosis | 2003

Q-VD-OPh, a broad spectrum caspase inhibitor with potent antiapoptotic properties.

Tina M. Caserta; A. N. Smith; Amy D. Gultice; Meredith A. Reedy; Thomas L. Brown

In recent years, several inhibitors that prevent caspase activation and apoptosis have emerged. At high doses, however, these inhibitors can have nonspecific effects and/or become cytotoxic. In this study, we determined the effectiveness of broad spectrum caspase inhibitors to prevent apoptosis. A carboxy terminal phenoxy group conjugated to the amino acids valine and aspartate (Q-VD-OPh) potently inhibited apoptosis. Q-VD-OPh was significantly more effective in preventing apoptosis than the widely used inhibitors, ZVAD-fmk and Boc-D-fmk, and was also equally effective in preventing apoptosis mediated by the three major apoptotic pathways, caspase 9/3, caspase 8/10, and caspase 12. In addition to the increased effectiveness, Q-VD-OPh was not toxic to cells even at extremely high concentrations. Our data indicate that the specificity, effectiveness, and reduced toxicity of caspase inhibitors can be significantly enhanced using carboxyterminal o-phenoxy groups and may have important uses in vivo.


Cancer Chemotherapy and Pharmacology | 2010

Progression and Treatment of HER2-Positive Breast Cancer

April Davoli; Barbara A. Hocevar; Thomas L. Brown

PurposeApproximately 20–30% of breast cancer tumors overexpress or amplify human epidermal growth factor receptor 2 (HER2). The role of this receptor in the progression of HER2+ breast cancer and resistance to certain anticancer monotherapies was investigated. The results of several pre-clinical and clinical trials, with the aim of determining the most safe and effective course of treatment for HER2+ breast cancer, were also thoroughly examined.MethodsA thorough search of databases including Pubmed, Springer, and The American Society of Clinical Oncology was performed, and pertinent studies were identified. The most relevant studies were preclinical, phase II, and III clinical trials identifying the function of the HER2 receptor in HER2+ breast cancer progression, as well as studies assessing the efficacy of monotherapy and combination therapy in the treatment of this aggressive form of cancer.ResultsThe HER2 receptor belongs to a family of receptors that consists of four cell-surface receptors (HER1-4) that share strong homology with the epidermal growth factor receptor (EGFR). All HER receptors interact with specific types of ligands to induce receptor activation, except for HER2, for which no known ligand has yet been identified. HER2 is activated by forming dimers with other HER receptors, and this results in a stronger and more prolonged signal transduction event. When expressed at normal levels, HER2 regulates cell growth, differentiation, and survival. However, under pathological conditions of HER2 overexpression, numerous HER2 heterodimers are formed resulting in aggressive tumor growth. Therefore, the prognosis associated with HER2-positive breast cancer is usually poor. A specific cohort of patients with breast cancer whose tumors test both hormone receptor (estrogen receptor [ER] and progesterone receptor [PR]) and HER2 positive have been found to be resistant to targeted hormone therapy. Studies investigating the etiology of this resistance have found that the cell membrane estrogen receptor communicates with HER2 in promoting the release of ER coactivators that cause the endocrine drug and selective estrogen receptor modulator, tamoxifen, to act as an agonist rather than an antagonist of the hormone estrogen. Thus, research has directed its inquiry toward the development of therapies specifically targeting HER2. The development of trastuzumab, a recombinant monoclonal antibody against HER2, initially proved to be a well-tolerated first line of treatment. However, in the long-term patients, trastuzumab was shown to develop resistance to this monotherapy. Therefore, research on HER2 positive breast cancer has focused on the study of different anti-HER2 combination therapies over the past decade.ConclusionsWhile the development and approval of the HER2-targeted recombinant monoclonal antibody trastuzumab (Herceptin) has been efficacious in slowing HER2 cancer progression, combining this and other anti-HER2 therapy with either chemotherapy or endocrine therapy has proven more effective in improving overall and progression free survival.


Apoptosis | 2006

Interferon-gamma sensitizes the human salivary gland cell line, HSG, to tumor necrosis factor-alpha induced activation of dual apoptotic pathways

Kashmira Kulkarni; Kaisa Selesniemi; Thomas L. Brown

Activated immune cells secrete proinflammatory cytokines such as tumor necrosis factor alpha (TNF-alpha), interferon–gamma (IFN-gamma) and Fas ligand (FasL) and these cytokines have been reported to induce apoptosis in numerous cell types. Apoptotic cell death has been associated with the progression of numerous autoimmune diseases. Proinflammatory cytokines are reportedly involved in apoptosis in the salivary glands of patients with Sjögren’s syndrome (SS); an autoimmune disorder characterized by the destruction of salivary and lachrymal glands. In this study, we used the HSG cell line to determine if exposure to proinflammatory cytokines induces apoptosis in human salivary gland cells. In addition, we identified the mediators controlling the apoptotic process in response to TNF alpha and IFN gamma. TNF-alpha and IFN-gamma induced apoptosis in HSG cells and resulted in the activation of caspase 8 and the “death receptor” pathway. We further determined that caspase 9 and the “mitochondrial” pathway was also activated. Induction of the intrinsic and extrinsic pathways in HSG cells resulted in substrate cleavage by effector caspases, in particular the cleavage of alpha II spectrin, an autoantigen in Sjögren’s syndrome. Our results suggest that HSG cells provide a model system to study processes regulating proinflammatory cytokine-induced apoptotic cell death.


Biology of Reproduction | 2006

Hypoxia Inhibits Differentiation of Lineage-Specific Rcho-1 Trophoblast Giant Cells

Amy D. Gultice; Kaisa Selesniemi; Thomas L. Brown

Abstract Defects in placental development lead to pregnancies at risk for miscarriage and intrauterine growth retardation and are associated with preeclampsia, a leading cause of maternal death and premature birth. In preeclampsia, impaired placental formation has been associated with alterations in a specific trophoblast lineage, the invasive trophoblast cells. In this study, an RT-PCR Trophoblast Gene Expression Profile previously developed by our laboratory was utilized to examine the lineage-specific gene expression of the rat Rcho-1 trophoblast cell line. Our results demonstrated that Rcho-1 cells represent an isolated, trophoblast population committed to the giant cell lineage. RT-PCR analysis revealed that undifferentiated Rcho-1 cells expressed trophoblast stem cell marker, Id2, and trophoblast giant cell markers. On differentiation, Rcho-1 cells downregulated Id2 and upregulated Csh1, a marker of the trophoblast giant cell lineage. Neither undifferentiated nor differentiated Rcho-1 cells expressed spongiotrophoblast marker Tpbpa or labyrinthine markers Esx1 and Tec. Differentiating Rcho-1 cells in hypoxia did not alter the expression of lineage-specific markers; however, hypoxia did inhibit the downregulation of the trophoblast stem cell marker Id2. Differentiation in hypoxia also blocked the induction of CSH1 protein. In addition, hypoxia inhibited stress fiber formation and abolished the induction of palladin, a protein associated with stress fiber formation and focal adhesions. Thus, Rcho-1 cells can be maintained as a proliferative, lineage-specific cell line that is committed to the trophoblast giant cell lineage on differentiation in both normoxic and hypoxic conditions; however, hypoxia does inhibit aspects of trophoblast giant cell differentiation at the molecular, morphological, and functional levels.


Apoptosis | 2003

Identification of the Primary Caspase 3 Cleavage Site in alpha II-Spectrin during Apoptosis

S. T. Williams; A. N. Smith; Carol D. Cianci; Jon S. Morrow; Thomas L. Brown

Alpha II-spectrin is one of the major proteins responsible for maintaining the cytoskeletal integrity of the cell. The caspase 3-mediated cleavage of alpha II-spectrin during apoptotic cell death may play an important role in altering membrane stability and the formation of apoptotic bodies. In this study, we identified the primary caspase 3 cleavage site in alpha II-spectrin. We found that the transcriptional inhibitor, actinomycin D, induced caspase 3 activation and that caspase 3 activation is coincident with the cleavage of alpha II-spectrin protein at a primary cleavage site. Deletion analysis and site directed mutagenesis identified the primary cleavage site in alpha II spectrin at amino acid 1185 (DETD). The primary caspase 3 cleavage site in alpha II spectrin is conserved in immature and mature B cells. Our results indicate that alpha II-spectrin is initally cleaved at a caspase 3 consensus site and this primary event likely alters the structural conformation of the protein exposing subsequent cleavage sites and altering cytoskeletal integrity. Identification of the primary cleavage site for caspase 3 may help to elucidate the role of alpha II-spectrin in membrane stability and apoptosis as well as provide new insights into alpha II-spectrin autoantibody formation associated with the autoimmune disease, Sjögrens syndrome.


Apoptosis | 2003

MTS-conjugated-antiactive caspase 3 antibodies inhibit actinomycin D-induced apoptosis.

Yang Zhao; Thomas L. Brown; Heinz Kohler; Sybille Müller

Caspase 3 is critically involved in the pathway of apoptosis. We have conjugated a MTS-transport-peptide to monoclonal and polyclonal anti-caspase-3 antibodies to suppress Actinomycin D-induced apoptosis in human lymphoma T cells. The advantage of using trans-membrane antibodies compared to conventional apoptosis inhibitors is their specific target recognition in the living cell and their lower toxicity compared to conventional apoptosis inhibitors. We could show that a MTS-transport-peptide modified monoclonal anti-caspase-3 antibody reduces Actinomycin D induced apoptosis, as shown by DNA ladder electrophoresis and cell death ELISA. These results indicate that antibodies have a therapeutic potential to inhibit apoptosis in a variety of diseases.


Cancer Growth and Metastasis | 2009

BITC Sensitizes Pancreatic Adenocarcinomas to TRAIL-Induced Apoptosis

Christina A. Wicker; Ravi P. Sahu; Kashmira Kulkarni-Datar; Sanjay K. Srivastava; Thomas L. Brown

Pancreatic adenocarcinoma is an aggressive cancer with a greater than 95% mortality rate and short survival after diagnosis. Chemotherapeutic resistance hinders successful treatment. This resistance is often associated with mutations in codon 12 of the K-Ras gene (K-Ras 12), which is present in over 90% of all pancreatic adenocarcinomas. Codon 12 mutations maintain Ras in a constitutively active state leading to continuous cellular proliferation. Our study determined if TRAIL resistance in pancreatic adenocarcinomas with K-Ras 12 mutations could be overcome by first sensitizing the cells with Benzyl isothiocyanate (BITC). BITC is a component of cruciferous vegetables and a cell cycle inhibitor. BxPC3, MiaPaCa2 and Panc-1 human pancreatic adenocarcinoma cell lines were examined for TRAIL resistance. Our studies show BITC induced TRAIL sensitization by dual activation of both the extrinsic and intrinsic apoptotic pathways.


Expert Opinion on Biological Therapy | 2005

TransMabs: cell-penetrating antibodies, the next generation

Sybille Müller; Yunfeng Zhao; Thomas L. Brown; Alton C. Morgan; Heinz Kohler

Intracellular proteins are becoming attractive targets in diagnosis and for therapy such as in signal pathways, on enzymes, transcription factors and structural proteins. Antibodies have been used therapeutically for extracellular pathogens and for targeting cell-surface antigens. Antibodies normally do not pass easily through intact cellular or subcellular membranes in living cells. Methods to shuttle antibodies into living cells are either labour-intensive and/or compromise the structural and functional integrity of the cell or require the integration of genes for heavy and light chain production through gene therapy approaches. A new technology platform, ‘SuperAntibody Technology’, enables antibodies to be shuttled into living cells without harming them. Such cell-penetrating antibodies open new diagnostic and therapeutic windows. The term ‘TransMabs’ has been coined for these antibodies. Proof of principle has been achieved with a 17-amino acid peptide with membrane translocating properties, conjugated with anti-caspase-3 antibody. Such a TransMab inhibits significantly invitro apoptosis-related events, such as caspase-3 activity, DNA fragmentation and spectrin cleavage. Anti-ca-spase-3 TransMab, therefore, could be utilised to inhibit apoptosis in a variety of diseases, such as Alzheimer’s, Huntington’s and P-arkinson’s. Unlike peptide inhibitors available at present, this TransMab is not expected to have invivo toxic side effects and can only target activated forms of the enzyme. This paper discusses the advantages and limitations of cell-penetrating antibodies (TransMabs) compared with existing small molecule drug development approaches.

Collaboration


Dive into the Thomas L. Brown's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chanel Keoni

Wright State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heinz Kohler

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge